US20220054484A1 - Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma - Google Patents

Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma Download PDF

Info

Publication number
US20220054484A1
US20220054484A1 US17/599,729 US202017599729A US2022054484A1 US 20220054484 A1 US20220054484 A1 US 20220054484A1 US 202017599729 A US202017599729 A US 202017599729A US 2022054484 A1 US2022054484 A1 US 2022054484A1
Authority
US
United States
Prior art keywords
fgfr3
fgfr
genetic alterations
fgfr2
fgfr genetic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/599,729
Other languages
English (en)
Inventor
Anjali Narayan AVADHANI
Anne Elizabeth O'HAGAN
Ademi Elena SANTIAGO-WALKER
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceutica NV
Janssen Pharmaceuticals Inc
Original Assignee
Janssen Pharmaceutica NV
Janssen Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Pharmaceutica NV, Janssen Pharmaceuticals Inc filed Critical Janssen Pharmaceutica NV
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Janssen Pharmaceuticals, Inc.
Assigned to Janssen Pharmaceuticals, Inc. reassignment Janssen Pharmaceuticals, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AVADHANI, ANJALI NARAYAN, SANTIAGO-WALKER, Ademi Elena, O'HAGAN, Anne Elizabeth
Publication of US20220054484A1 publication Critical patent/US20220054484A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • FGFR fibroblast growth factor receptor
  • methods of treating urothelial carcinoma in a patient comprising evaluating a biological sample from the patient for the presence of at least two fibroblast growth factor receptor (FGFR) genetic alterations and treating the patient with an FGFR inhibitor if the at least two fibroblast growth factor receptor (FGFR) genetic alterations are present in the sample. Also disclosed herein are methods of treating urothelial carcinoma in a patient harboring at least two fibroblast growth factor receptor (FGFR) genetic alterations comprising administering a FGFR inhibitor.
  • FGFR fibroblast growth factor receptor
  • FGFRs Fibroblast growth factor receptors
  • Described here are methods of treating urothelial carcinoma in a patient comprising, consisting of, or consisting essentially of: (a) evaluating a biological sample from the patient for the presence of at least two FGFR genetic alterations, wherein: (i) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (ii) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (iii) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (iv) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (v) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is
  • Also described herein are methods of treating urothelial carcinoma in a patient harboring at least two FGFR genetic alterations comprising, consisting of, or consisting essentially of administering a FGFR inhibitor to the patient, wherein: (a) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (b) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two
  • FGFR genetic alterations is an FGFR3 fusion; (c) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (d) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (e) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • the methods of treating urothelial carcinoma in a patient harboring at least two FGFR genetic alterations further comprise evaluating a biological sample from the patient for the presence of the at least two FGFR genetic alterations prior to administration of the FGFR inhibitor.
  • two or more of the at least two FGFR genetic alterations are FGFR2 fusions.
  • two or more FGFR genetic alterations comprise FGFR2-BICC1 and FGFR2-CASP7.
  • one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • two or more FGFR genetic alterations comprise FGFR2-CASP7 and FGFR3-BAIAP2L1; FGFR2-CASP7 and FGFR3-TACC3 V1; or FGFR2-CASP7 and FGFR3-TACC3 V3.
  • two or more of the at least two FGFR genetic alterations are FGFR3 mutations.
  • two or more FGFR genetic alterations comprise FGFR3 G370C and FGFR3 S249C; FGFR3 R248C and FGFR3 Y373C; or FGFR3 S249C and FGFR3 Y373C.
  • one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion.
  • two or more FGFR genetic alterations comprise FGFR3 G370C/FGFR2-BICC1; or FGFR3 S249C, FGFR3 Y373C, FGFR2-CASP7, FGFR3-BAIAP2L1, FGFR3-TACC3 V1 and FGFR3_TACC3 V3.
  • one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • the two or more FGFR genetic alterations are FGFR3 G370C and FGFR3-TACC3 V1; FGFR3 R248C and FGFR3-TACC3 V1; FGFR3 S249C and FGFR3-BAIAP2L1; FGFR3 R248C, FGFR3 5249 and FGFR3-TACC3 V1; or FGFR3 S249C, FGFR3 Y373C, FGFR2-CASP7, FGFR3-BAIAP2L1, FGFR3-TACC3 V1 and FGFR3-TACC3 V3.
  • the urothelial carcinoma is locally advanced or metastatic.
  • the biological sample is blood, lymph fluid, bone marrow, a solid tumor sample, or any combination thereof.
  • the FGFR inhibitor is erdafitinib.
  • erdafitinib is administered daily, in particular once daily.
  • erdafitinib is administered orally.
  • erdafitinib is administered orally on a continuous daily dosing schedule.
  • erdafitinib is administered orally at a dose of about 8 mg once daily.
  • erdafitinib is administered orally at a dose of about 8 mg once daily on a continuous daily dosing schedule.
  • the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 14 to 21 days after initiating treatment if: (a) the patient exhibits a serum phosphate (PO4) level that is less than about 5.5 mg/dL at 14-21 days after initiating treatment; and (b) administration of erdafitinib at 8 mg once daily resulted in no ocular disorder or (c) administration of erdafitinib at 8 mg once daily resulted in no Grade 2 or greater adverse reaction.
  • PO4 serum phosphate
  • erdafitinib is present in a solid dosage form.
  • the solid dosage form is a tablet.
  • FIG. 1 represents the study scheme for the Phase 2, multicenter, open-label study to evaluate the efficacy and safety of erdafitinib in subjects with metastatic or surgically unresectable urothelial cancer harboring selected FGFR (fibroblast growth factor receptor) genetic alterations (FGFR translocations or mutations).
  • FGFR fibroblast growth factor receptor
  • FIG. 2 shows patient responses to treatment with 8 mg per day continuous erdafitinib (Regimen 3) : Objective response rates (ORRs) among patient subgroups.
  • FIG. 3 which comprises FIGS. 3A-C , shows waterfall plots of reduction in the sum of target lesion diameters after treatment with erdafitinib. Reductions in patients treated with ( FIG. 3A ) 8 mg per day continuous erdafitinib (regimen 3), ( FIG. 3B ) 10 mg intermittent erdafitinib (regimen 1), and ( FIG. 3C ) 6 mg per day continuous erdafitinib (regimen 2) among all treated patients.
  • FIG. 4 is a swimmer plot of responses to treatment with erdafitinib among all patients treated with 8 mg per day continuous erdafitinib. Responses per investigator assessment
  • FIG. 5 which comprises FIGS. 5A-5B , depicts progression-free survival and overall survival among patients treated with 8 mg per day continuous erdafitinib (Regimen 3). Kaplan-Meier curve of ( FIG. 5A ) progression-free survival and ( FIG. 5B ) overall survival after treatment with 8 mg continuous erdafitinib.
  • FIG. 6 which comprises FIGS. 6A-6B , depicts overall survival among patients treated with 10 mg intermittent and 6 mg per day continuous erdafitinib.
  • each said step may also be considered an independent embodiment in itself, combinable with others.
  • transitional terms “comprising,” “consisting essentially of,” and “consisting” are intended to connote their generally in accepted meanings in the patent vernacular; that is, (i) “comprising,” which is synonymous with “including,” “containing,” or “characterized by,” is inclusive or open-ended and does not exclude additional, unrecited elements or method steps; (ii) “consisting of excludes any element, step, or ingredient not specified in the claim; and (iii) “consisting essentially of limits the scope of a claim to the specified materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention.
  • the basic and novel characteristics relates to the ability of the method to provide at least one of the benefits described herein, including but not limited to the ability to improve the survivability of the human population relative to the survivability of the comparative human population described elsewhere herein.
  • Embodiments described in terms of the phrase “comprising” (or its equivalents) also provide, as embodiments, those which are independently described in terms of “consisting of and “consisting essentially of ”
  • the term “about” signifies a variance of ⁇ 10% of the associated value, but additional embodiments include those where the variance may be ⁇ 5%, ⁇ 15%, ⁇ 20%, ⁇ 25%, or ⁇ 50%.
  • FGFR fibroblast growth factor receptor
  • FGFR3-TACC3 v1 fusion between genes encoding FGFR3 and transforming acidic coiled-coil containing protein 3 variant 1, also referred to herein as FGFR3-TACC3 V1
  • FGFR3-TACC3 v3 fusion between genes encoding FGFR3 and transforming acidic coiled-coil containing protein 3 variant 3, also referred to herein as FGFR3-TACC3_V2
  • FGFR3-BAIAP2L1 fusion between genes encoding FGFR3 and brain-specific angiogenesis inhibitor 1-associated protein 2-like protein 1
  • FGFR2-BICC1 fusion between genes encoding FGFR2 and bicaudal C homolog 1
  • FGFR2-CASP7 fusion between genes encoding FGFR2 and caspase 7).
  • patient is intended to mean any animal, in particular, mammals. Thus, the methods are applicable to human and nonhuman animals, although most preferably with humans.
  • patient and subject and “human” may be used interchangeably.
  • treat and “treatment” refer to the treatment of a patient afflicted with a pathological condition and refers to an effect that alleviates the condition by killing the cancerous cells, but also to an effect that results in the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, and cure of the condition.
  • Treatment as a prophylactic measure i.e., prophylaxis
  • cancer refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread).
  • co-administration encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.
  • pharmaceutical combination means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • fixed combination means that the active ingredients, e.g., erdafitinib and a co-agent, are both administered to a patient simultaneously in the form of a single unit or single dosage form.
  • non-fixed combination means that the active ingredients, e.g., erdafitinib and a co-agent, are administered to a patient as separate units or separate dosage forms, either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides safe and effective levels of the two active ingredients in the body of the human patient.
  • cocktail therapy e.g., the administration of three or more active ingredients.
  • continuous daily dosing schedule refers to the administration of a particular therapeutic agent without any drug holidays from the particular therapeutic agent.
  • a continuous daily dosing schedule of a particular therapeutic agent comprises administration of a particular therapeutic agent every day at roughly the same time each day.
  • progression-free survival is defined as the time from first dose to date of documented evidence of disease progression or death, whichever comes first.
  • duration of response is defined as the time from initial documentation of response to the date of documented evidence of disease progression or death.
  • all survival is defined as the time from first dose to the date of death. Data for patients who are alive or have unknown status is censored at the last date on which the patient is known to be alive.
  • placebo as used herein means administration of a pharmaceutical composition that does not include an FGFR inhibitor.
  • randomization refers to the time when the patient is confirmed eligible for the clinical trial and gets assigned to a treatment arm.
  • Bio samples refers to any sample for a patient in which cancerous cells can be obtained and detection of a FGFR genetic alteration is possible. Suitable biological samples include, but are not limited to, blood, lymph fluid, bone marrow, a solid tumor sample, or any combination thereof. In some embodiments, the biological sample can be formalin-fixed paraffin-embedded tissue (FFPET).
  • FFFPET formalin-fixed paraffin-embedded tissue
  • Described here are methods of treating urothelial carcinoma in a patient comprising, consisting of, or consisting essentially of: (a) evaluating a biological sample from the patient for the presence of at least two FGFR genetic alterations, wherein: (i) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (ii) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (iii) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (iv) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (v) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is
  • Also described herein are methods of treating urothelial carcinoma in a patient harboring at least two FGFR genetic alterations comprising, consisting of, or consisting essentially of administering a FGFR inhibitor to the patient, wherein: (a) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (b) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (c) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (d) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (e) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • FGF fibroblast growth factor
  • PTK protein tyrosine kinase receptors regulates a diverse array of physiologic functions including mitogenesis, wound healing, cell differentiation and angiogenesis, and development. Both normal and malignant cell growth as well as proliferation are affected by changes in local concentration of FGFs, extracellular signaling molecules which act as autocrine as well as paracrine factors. Autocrine FGF signaling may be particularly important in the progression of steroid hormone-dependent cancers to a hormone independent state. FGFs and their receptors are expressed at increased levels in several tissues and cell lines and overexpression is believed to contribute to the malignant phenotype.
  • FGFs and their receptors are expressed at increased levels in several tissues and cell lines and overexpression is believed to contribute to the malignant phenotype.
  • oncogenes are homologues of genes encoding growth factor receptors, and there is a potential for aberrant activation of FGF-dependent signaling in human pancreatic cancer (Knights et al., Pharmacology and Therapeutics 2010 125:1 (105-117); Korc M. et al Current Cancer Drug Targets 2009 9:5 (639-651)).
  • the two prototypic members are acidic fibroblast growth factor (aFGF or FGF1) and basic fibroblast growth factor (bFGF or FGF2), and to date, at least twenty distinct FGF family members have been identified.
  • the cellular response to FGFs is transmitted via four types of high affinity transmembrane protein tyrosine-kinase fibroblast growth factor receptors (FGFR) numbered 1 to 4 (FGFR1 to FGFR4).
  • FGFR high affinity transmembrane protein tyrosine-kinase fibroblast growth factor receptors
  • the urothelial carcinoma is susceptible to an FGFR2 genetic alteration or an FGFR3 genetic alteration. In further embodiments, the urothelial carcinoma is susceptible to at least two FGFR genetic alterations. In certain embodiments, the urothelial carcinoma is susceptible to at least two FGFR genetic alterations wherein: (i) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (ii) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (iii) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (iv) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (v) one or more of the at least two FGFR genetic alterations
  • FGFR genetic alteration refers to an alteration in the wild type FGFR gene, including, but not limited to, FGFR fusion genes, FGFR mutations, FGFR amplifications, or any combination thereof.
  • variant and “alteration” are used interchangeably herein.
  • the FGFR genetic alteration is an FGFR gene fusion.
  • FGFR fusion or “FGFR gene fusion” refers to a gene encoding a portion of FGFR (e.g., FGRF2 or FGFR3) and one of the herein disclosed fusion partners, or a portion thereof, created by a translocation between the two genes.
  • fusion and “translocation” are used interchangeable herein.
  • the presence of one or more of the following FGFR fusion genes in a biological sample from a patient can be determined using the disclosed methods: FGFR3-TACC3, FGFR3-BAIAP2L1, FGFR2-BICC1, FGFR2-CASP7, or any combination thereof.
  • FGFR-TACC3 is FGFR-TACC3 variant 1 (FGFR-TACC3 v1) or FGFR-TACC3 variant 3 (FGFR-TACC3 v3).
  • Table 1 provides the FGFR fusion genes and the FGFR and fusion partner exons that are fused. The sequences of the individual FGFR fusion genes are disclosed in Table 1.
  • FGFR genetic alterations include FGFR single nucleotide polymorphism (SNP).
  • SNP FGFR single nucleotide polymorphism
  • SNP refers to a FGFR2 or FGFR3 gene in which a single nucleotide differs among individuals.
  • the FGFR2 or FGFR3 genetic alteration is an FGFR3 gene mutation.
  • FGFR single nucleotide polymorphism refers to a FGFR3 gene in which a single nucleotide differs among individuals.
  • FGFR SNPs The presence of one or more of the following FGFR SNPs in a biological sample from a patient can be determined by methods known to those of ordinary skill in the art or methods disclosed in WO 2016/048833, FGFR3 R248C, FGFR3 S249C, FGFR3 G370C, FGFR3 Y373C, or any combination thereof.
  • the sequences of the FGFR SNPs are provided in Table 2.
  • the urothelial carcinoma is susceptible to at least two FGFR genetic alterations.
  • the FGFR alterations can be one or more FGFR fusion genes.
  • the FGFR alterations can be one or more FGFR mutations.
  • the FGFR alterations can be one or more FGFR amplifications.
  • a combination of the one or more FGFR alterations can be present in the biological sample from the patient.
  • one or more of the at least two FGFR genetic alterations is an FGFR mutation. In further embodiments, one or more of the at least two FGFR genetic alterations is an FGFR2 mutation. In still further embodiments, one or more of the at least two FGFR genetic alterations is an FGFR3 mutation. In some embodiments, the FGFR3 mutation is FGFR3 R248C, FGFR3 S249C, FGFR3 G370C, FGFR3 Y373C, or any combination thereof.
  • one or more of the at least two FGFR genetic alterations is an FGFR fusion. In further embodiments, one or more of the at least two FGFR genetic alterations is an FGFR3 fusion. In still further embodiments, the FGFR3 fusion is FGFR3-BAIAP2L1, FGFR3-TACC3 v1, FCFR3-TACC3 v3, or any combination thereof. In further embodiments, one or more of the at least two FGFR genetic alterations is an FGFR2 fusion. In still further embodiments, the FGFR2 fusion is FGFR2-BICC1, FGFR2-CASP7, or any combination thereof.
  • two or more of the at least two FGFR genetic alterations are FGFR2 fusions.
  • two or more FGFR genetic alterations comprise FGFR2-BICC1 and FGFR2-CASP7.
  • one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • two or more FGFR genetic alterations comprise FGFR2-CASP7 and FGFR3-BAIAP2L1; FGFR2-CASP7 and FGFR3-TACC3 V1; or FGFR2-CASP7 and FGFR3-TACC3 V3.
  • two or more of the at least two FGFR genetic alterations are FGFR3 mutations.
  • two or more FGFR genetic alterations comprise FGFR3 G370C and FGFR3 S249C; FGFR3 R248C and FGFR3 Y373C; or FGFR3 S249C and FGFR3 Y373C.
  • one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion.
  • two or more FGFR genetic alterations comprise FGFR3 G370C/FGFR2-BICC1; or FGFR3 S249C, FGFR3 Y373C, FGFR2-CASP7, FGFR3-BAIAP2L1, FGFR3-TACC3 V1 and FGFR3-TACC3 V3.
  • one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • two or more FGFR genetic alterations comprise FGFR3 G370C and FGFR3-TACC3 V1; FGFR3 R248C and FGFR3-TACC3 V1; FGFR3 S249C and FGFR3-BAIAP2L1; FGFR3 R248C, FGFR3 5249 and FGFR3-TACC3 V1; or FGFR3 S249C, FGFR3 Y373C, FGFR2-CASP7, FGFR3-BAIAP2L1, FGFR3-TACC3 V1 and FGFR3-TACC3 V3.
  • FGFR mutant gene panel includes one or more of the above listed FGFR mutants. In some embodiments, the FGFR mutant gene panel is dependent upon the patient's cancer type.
  • FGFR mutant panel that is used in the evaluating step of the disclosed methods is based, in part, on the patient's cancer type.
  • a suitable FGFR mutant gene panel can comprise FGFR3-TACC3 V1, FGFR3-TACC3 V3, FGFR3-BAIAP2L1, FGFR2-BICC1, FGFR2-CASP7, FGFR3 R248C, FGFR3 S249C, FGFR3 G370C, or FGFR3 Y373C, or any combination thereof.
  • FGFR inhibitors for use in the disclosed methods are provided herein.
  • the urothelial carcinoma patient can be treated with a FGFR inhibitor disclosed in U.S. Publication No. 2013/0072457 A1 (incorporated herein by reference), including any tautomeric or stereochemically isomeric form thereof, and a N-oxide thereof, a pharmaceutically acceptable salt thereof, or a solvate thereof (suitable R groups are also disclosed in U.S. Publication No. 2013/0072457 A1).
  • a FGFR inhibitor disclosed in U.S. Publication No. 2013/0072457 A1 (incorporated herein by reference), including any tautomeric or stereochemically isomeric form thereof, and a N-oxide thereof, a pharmaceutically acceptable salt thereof, or a solvate thereof (suitable R groups are also disclosed in U.S. Publication No. 2013/0072457 A1).
  • the patient may be treated with N-(3,5-dimethoxy-phenyl)-N′-(1-methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine (referred to herein “JNJ-42756493” or “JNJ493” or erdafitinib), including any tautomeric form thereof, N-oxides thereof, pharmaceutically acceptable salts thereof, or solvates thereof.
  • the FGFR inhibitor can be the compound of formula (I):
  • the pharmaceutically acceptable salt is a HCl salt.
  • erdafitinib base is used.
  • the urothelial carcinoma patient can be treated with a FGFR inhibitor wherein the FGFR inhibitor is N-[5-[2-(3,5-Dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-diemthylpiperazin-1-yl)benzamide (AZD4547), as described in Gavine, P. R., et al., AZD4547: An Orally Bioavailable, Potent, and Selective Inhibitor of the Fibroblast Growth Factor Receptor Tyrosine Kinase Family, Cancer Res. Apr. 15, 2012 72; 2045:
  • the FGFR inhibitor is N-[5-[2-(3,5-Dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-diemthylpiperazin-1-yl)benzamide (AZD4547), as described in Gavine, P. R., et al., AZD4547: An Or
  • any tautomeric or stereochemically isomeric form thereof including, when chemically possible, any tautomeric or stereochemically isomeric form thereof, and a N-oxide thereof, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • the urothelial carcinoma patient can be treated with a FGFR inhibitor wherein the FGFR inhibitor is 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-1- ⁇ 6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimid-4-yl ⁇ -methyl-urea (NVP-BGJ398) as described in Int'l Publ. No. WO2006/000420:
  • any tautomeric or stereochemically isomeric form thereof including, when chemically possible, any tautomeric or stereochemically isomeric form thereof, and a N-oxide thereof, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • the urothelial carcinoma patient can be treated with a FGFR inhibitor wherein the FGFR inhibitor is 4-amino-5-fluoro-3-[6-(4-methylpiperazin-1-yl)-1H-benzimidazol-2-yl]-1H-quinolin-2-one (dovitinib) as described in Int't Publ. No. WO2006/127926:
  • any tautomeric or stereochemically isomeric form thereof including, when chemically possible, any tautomeric or stereochemically isomeric form thereof, and a N-oxide thereof, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • the urothelial carcinoma patient can be treated with a FGFR inhibitor wherein the FGFR inhibitor is 6-(7-((1-Aminocyclopropyl)-methoxy)-6-methoxyquinolin-4-yloxy)-N-methyl-1-naphthamide (AL3810) (lucitanib; E-3810), as described in Bello, E. et al., E-3810 Is a Potent Dual Inhibitor of VEGFR and FGFR that Exerts Antitumor Activity in Multiple Preclinical Models, Cancer Res Feb. 15, 2011 71(A)1396-1405 and Int'l Publ. No. WO2008/112408:
  • any tautomeric or stereochemically isomeric form thereof including, when chemically possible, any tautomeric or stereochemically isomeric form thereof, and a N-oxide thereof, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • Additional suitable FGFR inhibitors include BAY1163877 (Bayer), BAY1179470 (Bayer), TAS-120 (Taiho), ARQ087 (ArQule), ASP5878 (Astellas), FF284 (Chugai), FP-1039 (GSK/FivePrime), Blueprint, LY-2874455 (Lilly), RG-7444 (Roche), or any combination thereof, including, when chemically possible, any tautomeric or stereochemical isomeric forms thereof, N-oxides thereof, pharmaceutically acceptable salts thereof, or solvates thereof.
  • the FGFR inhibitor generally, and erdafitinib more specifically, is administered as a pharmaceutically acceptable salt.
  • the FGFR inhibitor generally, is administered in base form.
  • the FGFR inhibitor generally, and erdafitinib more specifically, is administered as a pharmaceutically acceptable salt in an amount corresponding to 8 mg base equivalent or corresponding to 9 mg base equivalent.
  • the FGFR inhibitor generally, and erdafitinib more specifically, is administered in base form in an amount of 8 mg or 9 mg.
  • the salts can be prepared by for instance reacting the FGFR inhibitor generally, and erdafitinib more specifically, with an appropriate acid in an appropriate solvent.
  • Acid addition salts may be formed with acids, both inorganic and organic.
  • acid addition salts include salts formed with an acid selected from the group consisting of acetic, hydrochloric, hydriodic, phosphoric, nitric, sulphuric, citric, lactic, succinic, maleic, malic, isethionic, fumaric, benzenesulphonic, toluenesulphonic, methanesulphonic (mesylate), ethanesulphonic, naphthalenesulphonic, valeric, acetic, propanoic, butanoic, malonic, glucuronic and lactobionic acids.
  • Another group of acid addition salts includes salts formed from acetic, adipic, ascorbic, aspartic, citric, DL-Lactic, fumaric, gluconic, glucuronic, hippuric, hydrochloric, glutamic, DL-malic, methanesulphonic, sebacic, stearic, succinic and tartaric acids.
  • the FGFR inhibitor generally, and erdafitinib more specifically, is administered in the form of a solvate.
  • solvate means a physical association of erdafitinib with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • the term “solvate” is intended to encompass both solution-phase and isolatable solvates.
  • Non-limiting examples of solvents that may form solvates include water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid or ethanolamine and the like.
  • Solvates are well known in pharmaceutical chemistry. They can be important to the processes for the preparation of a substance (e.g. in relation to their purification, the storage of the substance (e.g. its stability) and the ease of handling of the substance and are often formed as part of the isolation or purification stages of a chemical synthesis.
  • a person skilled in the art can determine by means of standard and long used techniques whether a hydrate or other solvate has formed by the isolation conditions or purification conditions used to prepare a given compound. Examples of such techniques include thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), X-ray crystallography (e.g.
  • the compound may have one or more polymorph (crystalline) or amorphous forms.
  • the compounds include compounds with one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element.
  • a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T).
  • references to carbon and oxygen include within their scope respectively 12C, 13 C and 14 C and 16 O and 18 O.
  • the isotopes may be radioactive or nonradioactive.
  • the compounds contain no radioactive isotopes. Such compounds are preferred for therapeutic use.
  • the compound may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.
  • Described here are methods of treating urothelial carcinoma in a patient comprising, consisting of, or consisting essentially of: (a) evaluating a biological sample from the patient for the presence of at least two FGFR genetic alterations, wherein: (i) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (ii) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (iii) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (iv) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (v) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is
  • FGFR inhibitors for use in the treatment of urothelial carcinoma, said treatment comprising, consisting of, or consisting essentially of: (a) evaluating a biological sample from the patient for the presence of at least two FGFR genetic alterations, wherein: (i) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (ii) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (iii) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (iv) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (v) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR3 mutation
  • FGFR inhibitors in the manufacture of a medicament for the treatment of urothelial carcinoma, said treatment comprising, consisting of, or consisting essentially of: (a) evaluating a biological sample from the patient for the presence of at least two FGFR genetic alterations, wherein: (i) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (ii) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (iii) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (iv) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (v) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two
  • Also described herein are methods of treating urothelial carcinoma in a patient harboring at least two FGFR genetic alterations comprising, consisting of, or consisting essentially of administering a FGFR inhibitor to the patient, wherein: (a) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (b) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (c) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (d) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (e) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • FGFR inhibitors for use in the treatment of urothelial carcinoma in a patient harboring at least two FGFR genetic alterations, said treatment comprising, consisting of, or consisting essentially of administering a FGFR inhibitor to the patient, wherein: (a) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (b) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (c) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (d) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (e) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an
  • the urothelial carcinoma is locally advanced or metastatic.
  • the patient is a high-risk patient, in particular a high-risk patient with metastatic or surgically unresectable urothelial cancer, in particular metastatic or surgically unresectable urothelial cancer harboring select FGFR genetic alterations (FGFR translocations or mutations), in particular FGFR genetic alterations as defined herein.
  • a high-risk patient is a patient meeting one or more of the following criteria: age >75 years; ECOG PS 2; hemoglobin ⁇ 10 g/dL; visceral metastases, in particular of the liver, lung and/or bone; and 2 or 3 Bellmunt risk factors.
  • the hemoglobin level is measured in whole blood.
  • administration of the FGFR inhibitor provides improved anti-tumor activity as measured by objective response rate, progression-free survival, duration of response, or overall survival relative to a patient with urothelial carcinoma that is not receiving treatment with an FGFR inhibitor. In certain embodiments, administration of the FGFR inhibitor provides improved anti-tumor activity as measured by objective response rate or duration of response relative to a patient with urothelial carcinoma that is not receiving treatment with an FGFR inhibitor. In certain embodiments, administration of the FGFR inhibitor provides improved anti-tumor activity as measured by objective response rate relative to a patient with urothelial carcinoma that is not receiving treatment with an FGFR inhibitor.
  • administration of the FGFR inhibitor provides improved anti-tumor activity as measured by progression-free survival relative to a patient with urothelial carcinoma that is not receiving treatment with an FGFR inhibitor. In certain embodiments, administration of the FGFR inhibitor provides improved anti-tumor activity as measured by duration of response relative to a patient with urothelial carcinoma that is not receiving treatment with an FGFR inhibitor. In certain embodiments, administration of the FGFR inhibitor provides improved anti-tumor activity as measured by overall survival relative to a patient with urothelial carcinoma that is not receiving treatment with an FGFR inhibitor.
  • the improvement in anti-tumor activity is relative to treatment with placebo. In certain embodiments, the improvement in anti-tumor activity is relative to no treatment. In certain embodiments, the improvement in anti-tumor activity is relative to standard of care.
  • Measurable disease is defined by the presence of at least one measurable lesion.
  • administration of the FGFR inhibitor results in no more than a grade 2 adverse event. In other embodiments, administration of the FGFR inhibitor results in no more than a grade 3 adverse event. In some embodiments, administration of the FGFR inhibitor results in no more than a grade 4 adverse event.
  • the methods of treating urothelial carcinoma or the use in the treatment of urothelial carcinoma in a patient harboring at least two FGFR genetic alterations further comprise evaluating a biological sample from the patient for the presence of the at least two FGFR genetic alterations prior to administration of the FGFR inhibitor.
  • two or more of the at least two FGFR genetic alterations are FGFR2 fusions.
  • two or more FGFR genetic alterations comprise FGFR2-BICC1 and FGFR2-CASP7.
  • one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • two or more FGFR genetic alterations comprise FGFR2-CASP7 and FGFR3-BAIAP2L1; FGFR2-CASP7 and FGFR3-TACC3 V1; or FGFR2-CASP7 and FGFR3-TACC3 V3.
  • two or more of the at least two FGFR genetic alterations are FGFR3 mutations.
  • two or more FGFR genetic alterations comprise FGFR3 G370C and FGFR3 S249C; FGFR3 R248C and FGFR3 Y373C; or FGFR3 S249C and FGFR3 Y373C.
  • one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion.
  • two or more FGFR genetic alterations comprise FGFR3 G370C/FGFR2-BICC1; or FGFR3 S249C, FGFR3 Y373C, FGFR2-CASP7, FGFR3-BAIAP2L1, FGFR3-TACC3 V1 and FGFR3_TACC3 V3.
  • one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • two or more FGFR genetic alterations comprise FGFR3 G370C and FGFR3-TACC3 V1; FGFR3 R248C and FGFR3-TACC3 V1; FGFR3 S249C and FGFR3-BAIAP2L1; FGFR3 R248C, FGFR3 S249 and FGFR3-TACC3 V1; or FGFR3 S249C, FGFR3 Y373C, FGFR2-CASP7, FGFR3-BAIAP2L1, FGFR3-TACC3 V1 and FGFR3-TACC3 V3.
  • the at least two FGFR genetic alterations comprise FGFR3 G370C and FGFR3 S249C; or FGFR3 R248C and FGFR3 Y373C.
  • the at least two FGFR genetic alterations comprise FGFR3 G370C and FGFR2-BICC1; FGFR3 G370C and FGFR3-TACC3 V1; FGFR3 R248C and FGFR3-TACC3 V1; or FGFR3 R248C, FGFR3 S249 and FGFR3-TACC3 V1.
  • the at least two FGFR genetic alterations comprise FGFR3 G370C and FGFR3 S249C; FGFR3 R248C and FGFR3 Y373C; FGFR3 G370C and FGFR2-BICC1; FGFR3 G370C and FGFR3-TACC3 V1; FGFR3 R248C and FGFR3-TACC3 V1; or FGFR3 R248C, FGFR3 S249 and FGFR3-TACC3 V1.
  • Also described herein are methods of treating urothelial carcinoma in a patient harboring at least two FGFR genetic alterations comprising, consisting of, or consisting essentially of administering a FGFR inhibitor to the patient, wherein: (a) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (b) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (c) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (d) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (e) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion.
  • the methods of treating urothelial carcinoma in a patient harboring at least two FGFR genetic alterations further comprise evaluating a biological sample from the patient for the presence of the at least two FGFR genetic alterations prior to administration of the FGFR inhibitor.
  • the disclosed methods are suitable for treating cancer in a patient if at least two FGFR genetic alterations are present in a biological sample from the patient.
  • the FGFR genetic alterations can be one or more FGFR fusion genes.
  • the FGFR genetic alterations can be one or more FGFR mutations.
  • the FGFR genetic alterations can be one or more FGFR amplifications.
  • a combination of the one or more FGFR genetic alterations can be present in the biological sample from the patient.
  • the FGFR genetic alterations can be one or more FGFR fusion genes and one or more FGFR amplifications.
  • the FGFR genetic alterations can be one or more FGFR fusion genes and one or more FGFR mutations. In some embodiments, the FGFR alterations can be one or more FGFR mutations and one or more FGFR amplifications. In yet other embodiments, the FGFR alterations can be one or more FGFR fusion genes, mutations, and amplifications. Exemplary FGFR fusion genes are provided in Table 1 and include, but are not limited to, FGFR2-BICC1; FGFR2-CASP7; FGFR3-BAIAP2L1; FGFR3-TACC3 V1; FGFR3-TACC3 V3; or any combination thereof. Exemplary FGFR3 mutations are provided in Table 2 and include, but are not limited to, FGFR3 R248C, FGFR3 S249C, FGFR3 G370C, FGFR3 Y373C, or any combination thereof.
  • evaluating a biological sample for the presence of at least two FGFR genetic alterations are described in the methods section herein and in WO 2016/048833, which are incorporated herein in their entirety.
  • evaluating a biological sample for the presence of one or more FGFR variants can comprise any combination of the following steps: isolating RNA from the biological sample; synthesizing cDNA from the RNA; and amplifying the cDNA (preamplified or non-preamplified).
  • evaluating a biological sample for the presence of one or more FGFR variants can comprise: amplifying cDNA from the patient with a pair of primers that bind to and amplify one or more FGFR variants; and determining whether the one or more FGFR variants are present in the sample.
  • the cDNA can be pre-amplified.
  • the evaluating step can comprise isolating RNA from the sample, synthesizing cDNA from the isolated RNA, and pre-amplifying the cDNA.
  • Suitable primer pairs for performing an amplification step include, but are not limited to, those disclosed in WO 2016/048833, as exemplified below:
  • the presence of the at least two FGFR genetic alterations can be evaluated at any suitable time point including upon diagnosis, following tumor resection, following first-line therapy, during clinical treatment, or any combination thereof
  • a biological sample taken from a patient may be analyzed to determine whether a condition or disease, such as cancer, that the patient is or may be suffering from is one which is characterized by a genetic abnormality or abnormal protein expression which leads to up-regulation of the levels or activity of FGFR or to sensitization of a pathway to normal FGFR activity, or to upregulation of these growth factor signaling pathways such as growth factor ligand levels or growth factor ligand activity or to upregulation of a biochemical pathway downstream of FGFR activation.
  • Examples of such abnormalities that result in activation or sensitization of the FGFR signal include loss of, or inhibition of apoptotic pathways, up-regulation of the receptors or ligands, or presence of genetic alterations of the receptors or ligands e.g. PTK variants.
  • Tumors with genetic alterations of FGFR1, FGFR2 or FGFR3 or FGFR4 or up-regulation, in particular over-expression of FGFR1, or gain-of-function genetic alterations of FGFR2 or FGFR3 may be particularly sensitive to FGFR inhibitors.
  • the methods, compounds, and uses can further comprise evaluating the presence of at least two FGFR genetic alterations in the biological sample before the administering step.
  • the diagnostic tests and screens are typically conducted on a biological sample selected from tumor biopsy samples, blood samples (isolation and enrichment of shed tumor cells), stool biopsies, sputum, chromosome analysis, pleural fluid, peritoneal fluid, buccal spears, biopsy, circulating DNA, or urine.
  • the biological sample is blood, lymph fluid, bone marrow, a solid tumor sample, or any combination thereof.
  • the biological sample is a solid tumor sample.
  • Screening methods could include, but are not limited to, standard methods such as reverse-transcriptase polymerase chain reaction (RT-PCR) or in-situ hybridization such as fluorescence in situ hybridization (FISH).
  • RT-PCR reverse-transcriptase polymerase chain reaction
  • FISH fluorescence in situ hybridization
  • Identification of an individual carrying an FGFR genetic alteration may mean that the patient would be particularly suitable for treatment with erdafitinib. Tumors may preferentially be screened for presence of a FGFR genetic alteration prior to treatment.
  • the screening process will typically involve direct sequencing, oligonucleotide microarray analysis, or a mutant specific antibody.
  • diagnosis of tumor with such genetic alterations could be performed using techniques known to a person skilled in the art and as described herein such as RT-PCR and FISH.
  • mutant forms of, for example FGFR can be identified by direct sequencing of, for example, tumor biopsies using PCR and methods to sequence PCR products directly as hereinbefore described.
  • PCR and methods to sequence PCR products directly as hereinbefore described.
  • all such well-known techniques for detection of the over expression, activation or mutations of the aforementioned proteins could be applicable in the present case.
  • telomere length is assessed by telomere amplification of the cDNA by PCR.
  • Methods of PCR amplification, the selection of primers, and conditions for amplification, are known to a person skilled in the art. Nucleic acid manipulations and PCR are carried out by standard methods, as described for example in Ausubel, F. M. et al., eds. (2004) Current Protocols in Molecular Biology, John Wiley & Sons Inc., or Innis, M. A. et al., eds. (1990) PCR Protocols: a guide to methods and applications, Academic Press, San Diego.
  • in situ hybridization comprises the following major steps: (1) fixation of tissue to be analyzed; (2) prehybridization treatment of the sample to increase accessibility of target nucleic acid, and to reduce nonspecific binding; (3) hybridization of the mixture of nucleic acids to the nucleic acid in the biological structure or tissue; (4) post-hybridization washes to remove nucleic acid fragments not bound in the hybridization, and (5) detection of the hybridized nucleic acid fragments.
  • the probes used in such applications are typically labelled, for example, with radioisotopes or fluorescent reporters.
  • Preferred probes are sufficiently long, for example, from about 50, 100, or 200 nucleotides to about 1000 or more nucleotides, to enable specific hybridization with the target nucleic acid(s) under stringent conditions.
  • Standard methods for carrying out FISH are described in Ausubel, F. M. et al., eds. (2004) Current Protocols in Molecular Biology, John Wiley & Sons Inc and Fluorescence In Situ Hybridization: Technical Overview by John M. S. Bartlett in Molecular Diagnosis of Cancer, Methods and Protocols, 2nd ed.; ISBN: 1-59259-760-2; March 2004, pps. 077-088; Series: Methods in Molecular Medicine.
  • double-stranded cDNA is synthesized from total RNA Using a (dT)24 oligomer (SEQ ID NO: 38 : tttttttttttttttttttttttttttttttttttttttttttttttttttttttttttttttt) for priming first-strand cDNA synthesis, followed by second strand cDNA synthesis with random hexamer primers.
  • the double-stranded cDNA is used as a template for in vitro transcription of cRNA using biotinylated ribonucleotides.
  • cRNA is chemically fragmented according to protocols described by Affymetrix (Santa Clara, Calif., USA), and then hybridized overnight on Human Genome Arrays.
  • the protein products expressed from the mRNAs may be assayed by immunohistochemistry of tumor samples, solid phase immunoassay with microtitre plates, Western blotting, 2-dimensional SDS-polyacrylamide gel electrophoresis, ELISA, flow cytometry and other methods known in the art for detection of specific proteins. Detection methods would include the use of site-specific antibodies. The skilled person will recognize that all such well-known techniques for detection of upregulation of FGFR, and/or VEGFR, or detection of FGFR, and/or VEGFR variants or mutants could be applicable in the present case.
  • Abnormal levels of proteins such as FGFR can be measured using standard enzyme assays, for example, those assays described herein. Activation or overexpression could also be detected in a tissue sample, for example, a tumor tissue.
  • a tissue sample for example, a tumor tissue.
  • an assay such as that from Chemicon International. The tyrosine kinase of interest would be immunoprecipitated from the sample lysate and its activity measured.
  • Erdafitinib is in particular useful in treatment of a patient having at least two FGFR genetic alterations.
  • erdafitinib is useful in treating a patient having at least two FGFR genetic alterations, wherein: a) two or more of the at least two FGFR genetic alterations are FGFR2 fusions; (b) one or more of the at least two FGFR genetic alterations is an FGFR2 fusion and one or more of the at least two FGFR genetic alterations is an FGFR3 fusion; (c) two or more of the at least two FGFR genetic alterations are FGFR3 mutations; (d) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR2 fusion; or (e) one or more of the at least two FGFR genetic alterations is an FGFR3 mutation and one or more of the at least two FGFR genetic alterations is an FGFR3
  • the FGFR inhibitor generally, and erdafitinib more specifically, may be formulated into various pharmaceutical forms for administration purposes.
  • the pharmaceutical composition (e.g. formulation) comprises at least one active compound of the invention together with one or more pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art and optionally other therapeutic or prophylactic agents.
  • compositions an effective amount of the FGFR inhibitor generally and erdafitinib more specifically, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • a pharmaceutically acceptable carrier which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • the pharmaceutical compositions can be in any form suitable for oral, parenteral, topical, intranasal, ophthalmic, otic, rectal, intra-vaginal, or transdermal administration.
  • These pharmaceutical compositions are desirably in unitary dosage form suitable, preferably, for administration orally, rectally, percutaneously, or by parenteral injection.
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules and tablets.
  • compositions of the invention may include one or more pharmaceutically acceptable excipients (pharmaceutically acceptable carrier) such as disintegrants, diluents, fillers, binders, buffering agents, lubricants, glidants, thickening agents, sweetening agents, flavors, colorants, preservatives and the like.
  • pharmaceutically acceptable excipients such as disintegrants, diluents, fillers, binders, buffering agents, lubricants, glidants, thickening agents, sweetening agents, flavors, colorants, preservatives and the like.
  • Suitable disintegrants are those that have a large coefficient of expansion. Examples thereof are hydrophilic, insoluble or poorly water-soluble crosslinked polymers such as crospovidone (crosslinked polyvinylpyrrolidone) and croscarmellose sodium (crosslinked sodium carboxymethylcellulose).
  • the amount of disintegrant in the tablets according to the present invention may conveniently range from about 2.5 to about 15% w/w and preferably range from about 2.5 to 7% w/w, in particular range from about 2.5 to w/w. Because disintegrants by their nature yield sustained release formulations when employed in bulk, it is advantageous to dilute them with an inert substance called a diluent or filler.
  • a variety of materials may be used as diluents or fillers. Examples are lactose monohydrate, anhydrous lactose, sucrose, dextrose, mannitol, sorbitol, starch, cellulose (e.g. micro-crystalline cellulose (AvicelTM), silicified microcrystalline cellulose), dihydrated or anhydrous dibasic calcium phosphate, and others known in the art, and mixtures thereof (e.g. spray-dried mixture of lactose monohydrate (75%) with microcrystalline cellulose (25%) which is commercially available as MicrocelacTM). Preferred are microcrystalline cellulose and mannitol.
  • the total amount of diluent or filler in the pharmaceutical compositions of the present invention may conveniently range from about 20% to about 95% w/w and preferably ranges from about 55% to about 95% w/w, or from about 70% to about 95% w/w, or from about 80% to about 95% w/w, or from about 85% to about 95%.
  • Lubricants and glidants can be employed in the manufacture of certain dosage forms, and will usually be employed when producing tablets.
  • examples of lubricants and glidants are hydrogenated vegetable oils, e.g hydrogenated Cottonseed oil, magnesium stearate, stearic acid, sodium lauryl sulfate, magnesium lauryl sulfate, colloidal silica, colloidal anhydrous silica talc, mixtures thereof, and others known in the art.
  • interesting lubricants are magnesium stearate, and mixtures of magnesium stearate with colloidal silica, magnesium stearate being preferred.
  • a preferred glidant is colloidal anhydrous silica.
  • glidants generally comprise 0.2 to 7.0% w/w of the total composition weight, in particular 0.5 to 1.5% w/w, more in particular 1 to 1.5% w/w .
  • lubricants generally comprise 0.2 to 7.0% w/w of the total composition weight, in particular 0.2 to 2% w/w, or 0.5 to 2% w/w, or 0.5 to 1.75% w/w, or 0.5 to 1.5% w/w.
  • Binders can optionally be employed in the pharmaceutical compositions of the present invention.
  • Suitable binders are water-soluble polymers, such as alkylcelluloses such as methylcellulose; hydroxyalkylcelluloses such as hydroxymethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose and hydroxybutylcellulose; hydroxyalkyl alkylcelluloses such as hydroxyethyl methylcellulose and hydroxypropyl methylcellulose; carboxyalkylcelluloses such as carboxymethylcellulose; alkali metal salts of carboxyalkylcelluloses such as sodium carboxymethylcellulose; carboxyalkylalkylcelluloses such as carboxymethylethylcellulose; carboxyalkylcellulose esters; starches; pectines such as sodium carboxymethylamylopectine; chitin derivates such as chitosan; di-, oligo-and polysaccharides such as trehalose, cyclodextrins and derivatives thereof, alginic acid, alkali
  • Coloring agents and pigments include titanium dioxide and dyes suitable for food.
  • a coloring agent or a pigment is an optional ingredient in the formulation of the invention, but when used the coloring agent can be present in an amount up to 3.5% w/w based on the total composition weight.
  • Flavors are optional in the composition and may be chosen from synthetic flavor oils and flavoring aromatics or natural oils, extracts from plants leaves, flowers, fruits and so forth and combinations thereof. These may include cinnamon oil, oil of wintergreen, peppermint oils, bay oil, anise oil, eucalyptus, thyme oil. Also useful as flavors are vanilla, citrus oil, including lemon, orange, grape, lime and grapefruit, and fruit essences, including apple, banana, pear, peach, strawberry, raspberry, cherry, plum, pineapple, apricot and so forth. The amount of flavor may depend on a number of factors including the organoleptic effect desired. Generally, the flavor will be present in an amount from about 0% to about 3% (w/w).
  • Formaldehyde scavengers are compounds that are capable of absorbing formaldehyde. They include compounds comprising a nitrogen center that is reactive with formaldehyde, such as to form one or more reversible or irreversible bonds between the formaldehyde scavenger and formaldehyde.
  • the formaldehyde scavenger comprises one or more nitrogen atoms/centers that are reactive with formaldehyde to form a schiff base imine that is capable of subsequently binding with formaldehyde.
  • the formaldehyde scavenger comprises one or more nitrogen centers that are reactive with formaldehyde to form one or more 5-8 membered cyclic rings.
  • the formaldehyde scavenger preferably comprises one or more amine or amide groups.
  • the formaldehyde scavenger can be an amino acid, an amino sugar, an alpha amine compound, or a conjugate or derivative thereof, or a mixture thereof.
  • the formaldehyde scavenger may comprise two or more amines and/or amides.
  • Formaldehyde scavengers include, for example, glycine, alanine, serine, threonine, cysteine, valine, lecuine, isoleucine, methionine, phenylalanine, tyrosine, aspartic acid, glutamic acid, arginine, lysine, ornithine, citrulline, taurine pyrrolysine, meglumine, histidine, aspartame, proline, tryptophan, citrulline, pyrrolysine, asparagine, glutamine, or a conjugate or mixture thereof; or, whenever possible, pharmaceutically acceptable salts thereof.
  • the formaldehyde scavenger is meglumine or a pharmaceutically acceptable salt thereof, in particular meglumine base.
  • a formaldehyde scavenger in particular meglumine
  • erdafitinib a pharmaceutically acceptable salt thereof or a solvate thereof, in particular erdafitinib base
  • tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed.
  • the carrier will usually comprise sterile water, at least in large part, though other ingredients, to aid solubility for example, may be included.
  • injectable solutions for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution.
  • injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed.
  • the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not cause a significant deleterious effect to the skin.
  • Said additives may facilitate the administration to the skin and/or may be helpful for preparing the desired compositions.
  • These compositions may be administered in various ways, e.g., as a transdermal patch, as a spot-on, as an ointment. It is especially advantageous to formulate the aforementioned pharmaceutical compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used in the specification and claims herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • dosage unit forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, injectable solutions or suspensions, teaspoonfuls, tablespoonfuls and the like, and segregated multiples thereof.
  • Dosage unit form refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient, calculated to produce the desired therapeutic effect, in association with the required pharmaceutical carrier.
  • dosage unit forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, injectable solutions or suspensions, teaspoonfuls, tablespoonfuls and the like, and segregated multiples thereof.
  • Preferred forms are tablets and capsules.
  • the FGFR inhibitor, or erdafitinib specifically is present in a solid unit dosage form, and a solid unit dosage form suitable for oral administration.
  • the unit dosage form may contain about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mg of the FGFR inhibitor per unit dose form or an amount in a range bounded by two of these values, in particular 3, 4 or 5 mg per unit dose.
  • the pharmaceutical composition will preferably comprise from 0.05 to 99% by weight, more preferably from 0.1 to 70% by weight, even more preferably from 0.1 to 50% by weight of the compound of the present invention, and, from 1 to 99.95% by weight, more preferably from 30 to 99.9% by weight, even more preferably from 50 to 99.9% by weight of a pharmaceutically acceptable carrier, all percentages being based on the total weight of the composition.
  • Tablets or capsules of the present invention may further be film-coated e.g. to improve taste, to provide ease of swallowing and an elegant appearance.
  • Polymeric film-coating materials are known in the art.
  • Preferred film coatings are water-based film coatings opposed to solvent based film coatings because the latter may contain more traces of aldehydes.
  • a preferred film-coating material is Opadry® II aqueous film coating system, e.g. Opadry® II 85F, such as Opadry® II 85F92209.
  • Further preferred film coatings are water-based film coatings that protects from environmental moisture, such as Readilycoat® (e.g.
  • a preferred film-coating is Opadry® amb II, a high performance moisture barrier film coating which is a PVA-based immediate release system, without polyethylene glycol.
  • the film coat in terms of weight preferably accounts for about 4% (w/w) or less of the total tablet weight.
  • HPMC hypromellose
  • the pharmaceutical compositions as described herein in particular in the form of a capsule or a tablet, comprise from 0.5 mg to 20 mg base equivalent, or from 2 mg to 20 mg base equivalent, or from 0.5 mg to 12 mg base equivalent, or from 2 mg to 12 mg base equivalent, or from 2 mg to 10 mg base equivalent, or from 2 mg to 6 mg base equivalent, or 2 mg base equivalent, 3 mg base equivalent, 4 mg base equivalent, 5 mg base equivalent, 6 mg base equivalent, 7 mg base equivalent, 8 mg base equivalent, 9 mg base equivalent, 10 mg base equivalent, 11 mg base equivalent or 12 mg base equivalent of erdafitinib, a pharmaceutically acceptable salt thereof or a solvate thereof.
  • the pharmaceutical compositions as described herein comprise 3 mg base equivalent, 4 mg base equivalent or 5 mg base equivalent of erdafitinib, a pharmaceutically acceptable salt thereof or a solvate thereof.
  • the pharmaceutical compositions as described herein in particular in the form of a capsule or a tablet, comprise from 0.5 mg to 20 mg, or from 2 mg to 20 mg, or from 0.5 mg to 12 mg, or from 2 mg to 12 mg, or from 2 mg to 10 mg, or from 2 mg to 6 mg, or 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg or 12 mg of erdafitinib base.
  • the pharmaceutical compositions as described herein comprise 3 mg, 4 mg or 5 mg of erdafitinib base.
  • the pharmaceutical compositions as described herein comprise 3 mg, 4 mg or 5 mg of erdafitinib base and from about 0.5 to about 5% w/w, from about 0.5 to about 3% w/w, from about 0.5 to about 2% w/w, from about 0.5 to about 1.5% w/w, or from about 0.5 to about 1% w/w of a formaldehyde scavenger, in particular meglumine.
  • the pharmaceutical compositions as described herein comprise 3 mg, 4 mg or 5 mg of erdafitinib base and from about 0.5 to about 1.5% w/w or from about 0.5 to about 1% w/w of a formaldehyde scavenger, in particular meglumine.
  • more than one, e.g. two, pharmaceutical compositions as described herein can be administered in order to obtain a desired dose, e.g. a daily dose.
  • the amount of formaldehyde scavenger, in particular meglumine, in the pharmaceutical compositions according to the present invention may range from about 0.1 to about 10% w/w, about 0.1 to about 5% w/w, from about 0.1 to about 3% w/w, from about 0.1 to about 2% w/w, from about 0.1 to about 1.5% w/w, from about 0.1 to about 1% w/w, from about 0.5 to about 5% w/w, from about 0.5 to about 3% w/w, from about 0.5 to about 2% w/w, from about 0.5 to about 1.5% w/w, from about 0.5 to about 1% w/w.
  • All formulations for oral administration are in dosage form suitable for such administration.
  • the FGFR inhibitor generally, and erdafitinib specifically, is administered in an amount sufficient to exert its anti-tumor activity.
  • a therapeutically effective amount would be from 0.005 mg/kg to 100 mg/kg body weight, and in particular from 0.005 mg/kg to 10 mg/kg body weight.
  • Said sub-doses may be formulated as unit dosage forms, for example, containing 0.5 to 500 mg, in particular 1 mg to 500 mg, more in particular 10 mg to 500 mg of active ingredient per unit dosage form.
  • FGFR inhibitor in one aspect, described herein are methods of treating urothelial carcinoma or use for the treatment of urothelial carcinoma comprising, consisting of, or consisting essentially of administering a safe and effective amount of an FGFR inhibitor to a patient with urothelial carcinoma, wherein the FGFR inhibitor is administered orally.
  • the FGFR inhibitor generally, and erdafitinib specifically, is administered daily, in particular once daily.
  • the FGFR inhibitor generally, and erdafitinib specifically, is administered twice-a-day.
  • the FGFR inhibitor generally, and erdafitinib specifically, is administered three times a day.
  • the FGFR inhibitor generally, and erdafitinib specifically is administered four times a day. In some embodiments, the FGFR inhibitor generally, and erdafitinib specifically, is administered every other day. In some embodiments, the FGFR inhibitor generally, and erdafitinib specifically, is administered weekly. In some embodiments, the FGFR inhibitor generally, and erdafitinib specifically, is administered twice a week. In some embodiments, the FGFR inhibitor generally, and erdafitinib specifically, is administered every other week. In some embodiments, the FGFR inhibitor generally, and erdafitinib specifically, is administered orally on a continuous daily dosage schedule.
  • doses of the FGFR inhibitor, and erdafitinib specifically, employed for treatment of the diseases or conditions described herein in humans are typically in the range of about 1 to 20 mg per day.
  • the FGFR inhibitor, and erdafitinib specifically is administered orally to the human at a dose of about 1 mg per day, about 2 mg per day, about 3 mg per day, about 4 mg per day, about 5 mg per day, about 6 mg per day, about 7 mg per day, about 8 mg per day, about 9 mg per day, about 10 mg per day, about 11 mg per day, about 12 mg per day, about 13 mg per day, about 14 mg per day, about 15 mg per day, about 16 mg per day, about 17 mg per day, about 18 mg per day, about 19 mg per day or about 20 mg per day.
  • erdafitinib is administered orally at a dose of about 6 mg once daily.
  • erdafitinib is administered orally at a dose of about 8 mg once daily. In some embodiments, erdafitinib is administered orally at a dose of about 8 mg once daily on a continuous daily dosing schedule.
  • the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 14 to 21 days after initiating treatment if: (a) the patient exhibits a serum phosphate (PO 4 ) level that is less than about 5.5 mg/dL at 14-21 days after initiating treatment; and (b) administration of erdafitinib at 8 mg once daily resulted in no ocular disorder; or (c) administration of erdafitinib at 8 mg once daily resulted in no Grade 2 or greater adverse reaction.
  • PO 4 serum phosphate
  • the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 14 days after initiating treatment. In certain embodiments, the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 15 days after initiating treatment. In certain embodiments, the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 16 days after initiating treatment. In certain embodiments, the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 17 days after initiating treatment. In certain embodiments, the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 18 days after initiating treatment.
  • the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 19 days after initiating treatment. In certain embodiments, the dose of erdafitinib is increased from 8 mg once daily to 9 mg once daily at 20 days after initiating treatment.
  • erdafitinib is administered at a dose of 10 mg. In an embodiment, erdafitinib is administered at a dose of 10 mg intermittently. In an embodiment, erdafitinib is administered at a dose of 10 mg intermittently 7 days on/7 days off.
  • erdafitinib is administered at a dose of 8 mg, in particular 8 mg once daily. In an embodiment, erdafitinib is administered at a dose of 8 mg, in particular 8 mg once daily, with an option to uptitrate to 9 mg depending on serum phosphate levels (e.g. serum phosphate levels are ⁇ 5.5 mg/dL, or are ⁇ 7 mg/dL or range from and include 7 mg/dL to ⁇ 9 mg/dL or are ⁇ 9 mg/dL), and depending on treatment-related adverse events observed.
  • serum phosphate levels are ⁇ 5.5 mg/dL, or are ⁇ 7 mg/dL or range from and include 7 mg/dL to ⁇ 9 mg/dL or are ⁇ 9 mg/dL
  • the levels of serum phosphate for determining whether or not to up-titrate are measured on a treatment day during the first cycle of erdafitinib treatment, in particular on day 14 ⁇ 2 days, more in particular on day 14, of erdafitinib administration.
  • the treatment cycle as used herein is a 28-day cycle.
  • the desired dose is conveniently presented in a single dose or in divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • the FGFR inhibitor is conveniently presented in divided doses that are administered simultaneously (or over a short period of time) once a day.
  • the FGFR inhibitor is conveniently presented in divided doses that are administered in equal portions twice-a-day.
  • the FGFR inhibitor is conveniently presented in divided doses that are administered in equal portions three times a day.
  • the FGFR inhibitor is conveniently presented in divided doses that are administered in equal portions four times a day.
  • the desired dose may be delivered in 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fractional unit dosages throughout the course of the day, such that the total amount of FGFR inhibitor delivered by the fractional unit dosages over the course of the day provides the total daily dosage.
  • the amount of the FGFR inhibitor that is given to the human varies depending upon factors such as, but not limited to, condition and severity of the disease or condition, and the identity (e.g., weight) of the human, and the particular additional therapeutic agents that are administered (if applicable).
  • the at least one prior therapy for the treatment of urothelial carcinoma is platinum-containing chemotherapy.
  • the urothelial carcinoma progressed during or following at least one line of the platinum-containing chemotherapy.
  • the platinum-containing chemotherapy is neoadjuvant platinum-containing chemotherapy or adjuvant platinum-containing chemotherapy.
  • the urothelial carcinoma progressed during or within 12 months following at least one line of the neoadjuvant platinum-containing chemotherapy or adjuvant platinum-containing chemotherapy.
  • kits and articles of manufacture are also described.
  • kits include a package or container that is compartmentalized to receive one or more dosages of the pharmaceutical compositions disclosed herein.
  • Suitable containers include, for example, bottles.
  • the containers are formed from a variety of materials such as glass or plastic.
  • Packaging materials for use in packaging pharmaceutical products include, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252.
  • Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, bags, containers, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • a kit typically includes labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included.
  • a label is on or associated with the container.
  • a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • a label is used to indicate that the contents are to be used for a specific therapeutic application.
  • the label also indicates directions for use of the contents, such as in the methods described herein.
  • the pharmaceutical compositions are presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein.
  • the pack for example, contains metal or plastic foil, such as a blister pack.
  • the pack or dispenser device is accompanied by instructions for administration.
  • the pack or dispenser is also accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are also prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the nucleotide sequences for the FGFR fusion cDNA are provided in Table 5.
  • the underlined sequences correspond to either FGFR3 or FGFR2, the sequences in black represent the fusion partners and the sequence in italic fonts represent the intron sequence of the FGFR3 gene.
  • the study comprises a Screening Phase (molecular screening at any time prior to first dose and study screening within 30 days of first dose), a treatment phase, and a post-treatment follow-up phase.
  • the treatment phase comprises the period from first dose until the end-of-treatment visit.
  • the follow-up phase extends until the subject has died, withdraws consent, is lost to follow-up, or the end of study, whichever comes first.
  • Study treatment was administered on 28-day cycles. Prior to interim analysis 1, there were 2 treatment regimens. Patients were randomized 1:1 to 28-day cycles to the following 2 regimens until a regimen was selected for further study: Regimen 1 (10 mg once daily intermittent (7 days on/7 days); Regimen 2 (6 mg once daily continuous). Randomization was stratified according to performance status (0 to 1 vs. 2), hemoglobin value ( ⁇ 10 vs. ⁇ 10 g per dl), FGFR alteration type (mutation vs. fusion), prior treatment status (chemotherapy-resistant vs. chemotherapy na ⁇ ve), and disease distribution (presence or absence of visceral [liver, lung, bone] metastases). Starting dose selection was based on phase 1 efficacy and tolerability.
  • RNA from formalin-fixed, paraffin-embedded tumor samples were required to have at least 1 FGFR2/FGFR3 mutation or fusion per central lab testing of RNA from formalin-fixed, paraffin-embedded tumor samples, using a custom reverse transcriptase polymerase chain reaction assay.
  • Secondary end points include progression-free survival (PFS), response duration, Overall Survival, safety, response rate in biomarker-specific subgroups, and pharmacokinetics.
  • the median survival follow-up time was 22.9 months in the group receiving regimen 1 (interquartile range, 1.7+to 25.3+[95% CI, 20.5 to 24.5]) and 18.5 months (interquartile range, 0.4+to 21.6 [95% CI, 15.0 to 19.4) in the group receiving regimen 2.
  • the median numbers of cycles in regimens 1 and 2 were 5.0 (range, 1 to 25) and 4.5 (range, 1 to 22), respectively.
  • Median treatment durations were 4.4 and 3.9 months in regimens 1 and 2, respectively.
  • Chemotherapy-resistant patients were those who had progressed during or following ⁇ 1 line of prior systemic chemotherapy or within 12 months of adjuvant or neoadjuvant chemotherapy.
  • Chemotherapy-na ⁇ ve patients were those who were ineligible for cisplatin. Ineligibility for cisplatin was based on impaired renal function defined as 1) glomerular filtration rate ⁇ 60 mL/min/1.73 m2 by 24-hour urine measurement; 2) calculated by Cockcroft-Gault; or 3) grade 2 or higher peripheral neuropathy (CTCAE version 4.0).
  • Patients could have more than 1 FGFR alteration.
  • ORR The confirmed ORR (40.4%, with a two-sided 95% CI of 30.7% to 50.1%) per investigator assessment and time to response among patients treated with regimen 3 are presented in Table 7. Because lower boundary of the confidence interval was >25%, the primary end point was achieved. An additional 39 (39%) patients had stable disease for ⁇ 1 disease evaluation assessment (>36 days). Two patients had no postbaseline disease evaluations. ORRs were similar regardless of prior chemotherapy, number of prior treatment lines, presence of visceral metastases, or baseline characteristics such as age, sex, hemoglobin level, or renal function (Table 7, FIG. 2 ).
  • 87 patients had disease that had progressed on or after at least one prior chemotherapy (chemotherapy-relapsed/refractory disease) and that had at least 1 of the following gene alterations: FGFR3 gene mutations (R248C, S249C, G370C, Y373C) or FGFR gene fusions (FGFR3-TACC3, FGFR3-BAIAP2L1, FGFR2-BICC1, FGFR2-CASP7), as determined by a clinical trial assay performed at a central laboratory (Table 6).
  • the median age was 67 years (range: 36 to 87 years), 79% were male, and 74% were Caucasian.
  • FIG. 5A Median progression-free survival per investigator assessment at median follow-up of 11.2 months in patients receiving regimen 3 is presented in FIG. 5A . Progression-free survival rate (95% CI) at 12 months was 19% (11% to 29%). Median overall survival at median 11.0 months' follow-up for survival is presented in FIG. 5B . Survival rate at 12 months was 55% (43% to 66%).
  • Prophylactic measures were taken to minimize risk of common adverse events related to FGFR inhibition.
  • a low-phosphate diet was recommended for all patients (600 to 800 mg of dietary phosphate intake per day).
  • the application of alcohol-free emollient moisturizing cream and avoidance of unnecessary exposure to sunlight, soap, perfumed products, and hot baths was recommended. Patients were asked to keep their fingers and toes clean and nails trimmed to reduce risk of nail effects.
  • Treatment-related adverse events that were considered of special interest/clinical importance were hyperphosphatemia, skin effects, nail effects, and eye disorders, including central serous retinopathy (CSR) and other non-CSR ocular events (Table 18).
  • CSR central serous retinopathy
  • Treatment-related hyperphosphatemia and effects on the skin and on the nails were reported in 73%, 49%, and 52%, respectively, of patients treated with 8 mg per day continuous erdafitinib. Most events were mild to moderate. In this group, the most common treatment-related effects on the skin were dry skin (32%) and hand-foot syndrome (22%), and the most common treatment-related nail effects were nail dystrophy and onycholysis in 16% of patients each.
  • Hyperphosphatemia the most common treatment-related adverse event (Table 16, 14, 16), was managed by dose interruption (23%), dose reduction (9%), and treatment with phosphate binders when medically warranted. Phosphate elevation typically peaked 6 weeks after erdafitinib initiation and normalized by cycle 5. One patient discontinued treatment due to grade 1 hyperphosphatemia. Dry skin was managed with additional topical ointments such as ammonium lactate, salicylic acid, or zinc oxide creams. Nail effects were managed with topical nail strengthener, and antibiotics or silver nitrate were applied in severe cases.
  • median progression-free and overall survival were 5.5 months ( FIG. 5A ) and 13.8 months ( FIG. 5B ), respectively, including patients with visceral metastases and poor kidney function who had progressed on or after multiple lines of therapy.
  • 13 patients continued treatment beyond progression which was either limited progression in a target lesion or appearance of a small new lesion while the patient was assessed to have ongoing clinical benefit.
  • the safety profile allowed 8 mg continuous daily dosing, with uptitration to 9 mg daily dosing guided by serum phosphate levels.
  • pan-FGFR inhibitor erdafitinib had measurable benefit in patients with advance urothelial carcinoma with FGFR alterations.
  • Erdafitinib increased serum phosphate level as a consequence of FGFR inhibition.
  • Erdafitinib should be increased to the maximum recommended dose to achieve target serum phosphate levels of 5.5-7.0 mg/dL in early cycles with continuous daily dosing
  • phosphate binders were permitted. Avoid concomitant use with agents that can alter serum phosphate levels before the initial dose increase period based on serum phosphate levels.
  • the mean (coefficient of variation [CV %]) erdafitinib steady-state maximum observed plasma concentration (Cmax), area under the curve (AUCtau), and minimum observed plasma concentration (Cmin) were 1399 ng/mL (51%), 29268 ng ⁇ h/mL (60%), and 936 ng/mL (65%), respectively.
  • erdafitinib exposure increases proportionally across the dose range of 0.5 to 12 mg (0.06 to 1.3 times the maximum approved recommended dose). Steady state was achieved after 2 weeks with once daily dosing and the mean accumulation ratio was 4-fold.
  • the mean apparent volume of distribution of erdafitinib was 29 L in patients.
  • Erdafitinib protein binding was 99.8% in patients, primarily to alpha-1-acid glycoprotein.
  • the mean total apparent clearance (CL/F) of erdafitinib was 0.362 L/h in patients.
  • the mean effective half-life of erdafitinib was 59 hours in patients.
  • Erdafitinib is primarily metabolized by CYP2C9 and CYP3A4.
  • the contribution of CYP2C9 and CYP3A4 in the total clearance of erdafitinib is estimated to be 39% and 20% respectively.
  • Unchanged erdafitinib was the major drug-related moiety in plasma, there were no circulating metabolites.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Toxicology (AREA)
  • Hospice & Palliative Care (AREA)
  • Wood Science & Technology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
US17/599,729 2019-03-29 2020-03-27 Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma Pending US20220054484A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP19166428 2019-03-29
EP19166428.3 2019-03-29
EP19188971 2019-07-30
EP19188971.6 2019-07-30
PCT/US2020/025166 WO2020205493A1 (en) 2019-03-29 2020-03-27 Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma

Publications (1)

Publication Number Publication Date
US20220054484A1 true US20220054484A1 (en) 2022-02-24

Family

ID=70296138

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/599,729 Pending US20220054484A1 (en) 2019-03-29 2020-03-27 Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma

Country Status (14)

Country Link
US (1) US20220054484A1 (ar)
EP (1) EP3946341A1 (ar)
JP (1) JP2022527189A (ar)
KR (1) KR20210145211A (ar)
CN (1) CN113645974A (ar)
AU (1) AU2020253827A1 (ar)
BR (1) BR112021019446A2 (ar)
CA (1) CA3130460A1 (ar)
IL (1) IL286729A (ar)
JO (1) JOP20210266A1 (ar)
MA (1) MA55522A (ar)
MX (1) MX2021011926A (ar)
SG (1) SG11202109902SA (ar)
WO (1) WO2020205493A1 (ar)

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US6218529B1 (en) 1995-07-31 2001-04-17 Urocor, Inc. Biomarkers and targets for diagnosis, prognosis and management of prostate, breast and bladder cancer
US5882864A (en) 1995-07-31 1999-03-16 Urocor Inc. Biomarkers and targets for diagnosis, prognosis and management of prostate disease
GB0512324D0 (en) 2005-06-16 2005-07-27 Novartis Ag Organic compounds
WO2006127926A2 (en) 2005-05-23 2006-11-30 Novartis Ag Crystalline and other forms of 4-amino-5-fluoro-3-[6-(4-methylpiperazin-1-yl)-1h-benzimidazol-2-yl]-1h-quinolin-2-one lactic acid salts
US8163923B2 (en) 2007-03-14 2012-04-24 Advenchen Laboratories, Llc Spiro substituted compounds as angiogenesis inhibitors
GB201007286D0 (en) 2010-04-30 2010-06-16 Astex Therapeutics Ltd New compounds
LT3198033T (lt) 2014-09-26 2022-05-10 Janssen Pharmaceutica Nv Fgfr mutantinių genų rinkinių panaudojimas identifikuojant vėžiu sergančius pacientus, kurie reaguos į gydymą fgfr inhibitoriumi
AU2016341445B2 (en) * 2015-10-23 2020-08-27 Array Biopharma, Inc. 2-aryl- and 2-heteroaryl-substituted 2-pyridazin-3(2H)-one compounds as inhibitors of FGFR tyrosine kinases
JOP20190190A1 (ar) * 2017-02-06 2019-08-04 Janssen Pharmaceutica Nv معالجة سرطان
JOP20190280A1 (ar) * 2017-06-02 2019-12-02 Janssen Pharmaceutica Nv مثبطات fgfr2 لعلاج سرطان الأوعية الصفراوية

Also Published As

Publication number Publication date
AU2020253827A1 (en) 2021-09-30
EP3946341A1 (en) 2022-02-09
MA55522A (fr) 2022-02-09
JOP20210266A1 (ar) 2023-01-30
MX2021011926A (es) 2021-11-03
WO2020205493A1 (en) 2020-10-08
IL286729A (en) 2021-10-31
KR20210145211A (ko) 2021-12-01
CN113645974A (zh) 2021-11-12
BR112021019446A2 (pt) 2021-11-30
JP2022527189A (ja) 2022-05-31
SG11202109902SA (en) 2021-10-28
CA3130460A1 (en) 2020-10-08

Similar Documents

Publication Publication Date Title
TWI798199B (zh) 癌症治療
US20220110935A1 (en) Cancer Treatment
US20220175768A1 (en) Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma
US20230110113A1 (en) Fgfr tyrosine kinase inhibitors for the treatment of high-risk non-muscle invasive bladder cancer
AU2020223467A1 (en) Cancer treatment
US20220054484A1 (en) Fgfr tyrosine kinase inhibitors for the treatment of urothelial carcinoma
CN117320724A (zh) 用于治疗晚期实体瘤的fgfr酪氨酸激酶抑制剂
WO2022243467A1 (en) Fgfr tyrosine kinase inhibitors for the treatment of advanced solid tumors

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JANSSEN PHARMACEUTICALS, INC.;REEL/FRAME:058403/0100

Effective date: 20200326

Owner name: JANSSEN PHARMACEUTICALS, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AVADHANI, ANJALI NARAYAN;O'HAGAN, ANNE ELIZABETH;SANTIAGO-WALKER, ADEMI ELENA;SIGNING DATES FROM 20211006 TO 20211018;REEL/FRAME:058403/0067